1
|
Shi Y, Wen T, Zhao F, Hu J. Bacteriostasis of nisin against planktonic and biofilm bacteria: Its mechanism and application. J Food Sci 2024; 89:1894-1916. [PMID: 38477236 DOI: 10.1111/1750-3841.17001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/27/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024]
Abstract
Food safety incidents caused by bacterial contamination have always been one of the public safety issues of social concern. Planktonic cells, viable but non-culturable (VBNC) cells, and biofilm cells of bacteria can coexist in food or food processing, posing more serious challenges to public health and safety by increasing bacterial survival and difficulty in detection. As a non-toxic, no side effect, and highly effective bacteriostatic substance, nisin has received wide attention from researchers. In this review, we summarized the species and biosynthesis of nisin, the effects of nisin alone or in combination with other treatments on planktonic and biofilm cells, and its applications in the fields of food, feed, and medicine by consulting numerous studies. Meanwhile, the mechanism of nisin on planktonic and biofilm cells was proposed based on existing researches. Nisin not only has antibacterial activity against most G+ bacteria but also exhibits a bacteriostatic effect on G- bacteria when combined with other antibacterial treatments. In addition to planktonic cells, nisin also has significant effects on bacterial cells in biofilms by changing the thickness, density, and composition of biofilms. Based on the three action processes of nisin on biofilms, we summarized the changes of bacteria in biofilms, including the causes of bacterial death and the formation of the VBNC state. We consider that research on the relationship between nisin and VBNC state should be strengthened.
Collapse
Affiliation(s)
- Ying Shi
- College of Food Science and Engineering, Jilin University, Changchun, P. R. China
| | - Tao Wen
- College of Food Science and Engineering, Jilin University, Changchun, P. R. China
| | - Feng Zhao
- College of Food Science and Engineering, Jilin University, Changchun, P. R. China
| | - Jia Hu
- College of Food Science and Engineering, Jilin University, Changchun, P. R. China
| |
Collapse
|
2
|
Lavekar AG, Thakare R, Saima, Equbal D, Chopra S, Sinha AK. Indole-based aryl sulfides target the cell wall of Staphylococcus aureus without detectable resistance. Drug Dev Res 2024; 85:e22123. [PMID: 37840429 DOI: 10.1002/ddr.22123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/05/2023] [Accepted: 10/02/2023] [Indexed: 10/17/2023]
Abstract
Sulfur-containing classes of the scaffold "Arylthioindoles" have been evaluated for antibacterial activity; they demonstrated excellent potency against methicillin-resistant Staphylococcus aureus (MRSA) as well as against vancomycin-resistant strains and a panel of clinical isolates of resistant strains. In this study, we have elucidated the mechanism of action of lead compounds, wherein they target the cell wall of S. aureus. Further, S. aureus failed to develop resistance against two lead compounds tested in a serial passage experiment in the presence of the compounds over a period of 40 days. Both the compounds demonstrated comparable in vivo efficacy with vancomycin in a neutropenic mice thigh infection model. The results of these antibacterial activities emphasize the excellent potential of thioethers for developing novel antibiotics and may fill in as a target for the adjustment of accessible molecules to develop new powerful antibacterial agents with fewer side effects.
Collapse
Affiliation(s)
- Aditya G Lavekar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Ritesh Thakare
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Department of Cell and Cancer Biology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Saima
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- School of Advanced Chemical Sciences, Solan, Himachal Pradesh, India
| | - Danish Equbal
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Sidharth Chopra
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Arun K Sinha
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
- Ranchi University, Ranchi, Jharkhand, India
| |
Collapse
|
3
|
Panina IS, Balandin SV, Tsarev AV, Chugunov AO, Tagaev AA, Finkina EI, Antoshina DV, Sheremeteva EV, Paramonov AS, Rickmeyer J, Bierbaum G, Efremov RG, Shenkarev ZO, Ovchinnikova TV. Specific Binding of the α-Component of the Lantibiotic Lichenicidin to the Peptidoglycan Precursor Lipid II Predetermines Its Antimicrobial Activity. Int J Mol Sci 2023; 24:ijms24021332. [PMID: 36674846 PMCID: PMC9863751 DOI: 10.3390/ijms24021332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/21/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
To date, a number of lantibiotics have been shown to use lipid II-a highly conserved peptidoglycan precursor in the cytoplasmic membrane of bacteria-as their molecular target. The α-component (Lchα) of the two-component lantibiotic lichenicidin, previously isolated from the Bacillus licheniformis VK21 strain, seems to contain two putative lipid II binding sites in its N-terminal and C-terminal domains. Using NMR spectroscopy in DPC micelles, we obtained convincing evidence that the C-terminal mersacidin-like site is involved in the interaction with lipid II. These data were confirmed by the MD simulations. The contact area of lipid II includes pyrophosphate and disaccharide residues along with the first isoprene units of bactoprenol. MD also showed the potential for the formation of a stable N-terminal nisin-like complex; however, the conditions necessary for its implementation in vitro remain unknown. Overall, our results clarify the picture of two component lantibiotics mechanism of antimicrobial action.
Collapse
Affiliation(s)
- Irina S. Panina
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Sergey V. Balandin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Correspondence: ; Tel.: +7-495-335-0900
| | - Andrey V. Tsarev
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| | - Anton O. Chugunov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| | - Andrey A. Tagaev
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ekaterina I. Finkina
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Daria V. Antoshina
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Elvira V. Sheremeteva
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Alexander S. Paramonov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Jasmin Rickmeyer
- Institute of Medical Microbiology, Immunology and Parasitology, Medical Faculty, University of Bonn, 53117 Bonn, Germany
| | - Gabriele Bierbaum
- Institute of Medical Microbiology, Immunology and Parasitology, Medical Faculty, University of Bonn, 53117 Bonn, Germany
| | - Roman G. Efremov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- Department of Applied Mathematics, National Research University Higher School of Economics, 101000 Moscow, Russia
| | - Zakhar O. Shenkarev
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
| | - Tatiana V. Ovchinnikova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- Department of Bioorganic Chemistry, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
4
|
Thakur R, Suri CR, Kaur IP, Rishi P. Review. Crit Rev Ther Drug Carrier Syst 2022; 40:49-100. [DOI: 10.1615/critrevtherdrugcarriersyst.2022040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
5
|
Effects of sub-lethal doses of nisin on the virulence of Salmonella enterica in Galleria mellonella larvae. Res Microbiol 2021; 172:103836. [PMID: 34029676 DOI: 10.1016/j.resmic.2021.103836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/27/2021] [Accepted: 04/23/2021] [Indexed: 11/21/2022]
Abstract
Salmonella enterica is a pathogen that induces self-limiting gastroenteritis and is of worldwide concern. Nisin, an antimicrobial peptide, has emerged as an alternative for the control of microbial growth but its effect on the virulence of pathogenic bacteria is not yet well-explored. This work aimed to evaluate the virulence of S. enterica in the presence of sub-inhibitory nisin using the experimental model Galleria mellonella. Sub-inhibitory concentrations of nisin of 11.72 and 46.88 μM did not affect the cellular viability of S. enterica but promoted changes in gene expression within 1 h of treatment, with increases of up to 3-fold of pagC, 1.8-fold of invA and 2.3-fold of invF. Larvae of G. mellonella inoculated with S. enterica combined with nisin at 46.88 μM presented mortality, and TL50 noticeably increased to 50% and 80% at 24 and 48 h post-infection, respectively. Defence responses, such as melanisation, nodulation, pseudopodia, immune response, and expression of defence proteins of the larvae G. mellonella were enhanced when the treatments with S. enterica were combined with 11.72 or 46.88 μM nisin. These results show an increase in virulence of S. enterica by sub-MIC concentration of nisin that needs to be explored.
Collapse
|
6
|
Małaczewska J, Kaczorek-Łukowska E. Nisin-A lantibiotic with immunomodulatory properties: A review. Peptides 2021; 137:170479. [PMID: 33359393 DOI: 10.1016/j.peptides.2020.170479] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
Nisin, a member of class I bacteriocins known as lantibiotics, is produced by the lactic acid bacterium Lactococcus lactis and is characterized by a wide spectrum of antibacterial activity against gram-positive bacteria. This characteristic in conjunction with its low toxicity and safety of use in food has contributed to the worldwide success of nisin as a natural food preservative. This lantibiotic has attracted interest as a potential natural therapeutic agent for the control of bacterial infections. However, similar to other antimicrobial peptides of natural origin, the spectrum of biological activity of nisin surpasses its antibacterial properties, encompassing interesting and incompletely understood immunotropic characteristics. This paper is a systematic review of the current information about the potential immunomodulatory properties of nisin based on in vitro and in vivo studies in various experimental models. We also discuss the effect of potentially probiotic, nisin-producing L. lactis strains on the immune system of animals.
Collapse
Affiliation(s)
- Joanna Małaczewska
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Street 13, 10-718 Olsztyn, Poland.
| | - Edyta Kaczorek-Łukowska
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Street 13, 10-718 Olsztyn, Poland
| |
Collapse
|
7
|
Kaur A, Soni SK, Vij S, Rishi P. Cocktail of carbohydrases from Aspergillus niger: an economical and eco-friendly option for biofilm clearance from biopolymer surfaces. AMB Express 2021; 11:22. [PMID: 33538938 PMCID: PMC7862497 DOI: 10.1186/s13568-021-01183-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 01/18/2021] [Indexed: 11/25/2022] Open
Abstract
Biofilm formation on both biotic and abiotic surfaces accounts for a major factor in spread of antimicrobial resistance. Due to their ubiquitous nature, biofilms are of great concern for environment as well as human health. In the present study, an integrated process for the co-production of a cocktail of carbohydrases from a natural variant of Aspergillus niger was designed. The enzyme cocktail was found to have a noteworthy potential to eradicate/disperse the biofilms of selected pathogens. For application of enzymes as an antibiofilm agent, the enzyme productivities were enhanced by statistical modelling using response surface methodology (RSM). The antibiofilm potential of the enzyme cocktail was studied in terms of (i) in vitro cell dispersal assay (ii) release of reducing sugars from the biofilm polysaccharides (iii) the effect of enzyme treatment on biofilm cells and architecture by confocal laser scanning microscopy (CLSM). Potential of the enzyme cocktail to disrupt/disperse the biofilm of selected pathogens from biopolymer surfaces was also assessed by field emission scanning electron microscopy (FESEM) analysis. Further, their usage in conjunction with antibiotics was assessed and it was inferred from the results that the use of enzyme cocktail augmented the efficacy of the antibiotics. The study thus provides promising insights into the prospect of using multiple carbohydrases for management of heterogeneous biofilms formed in natural and clinical settings.
Collapse
Affiliation(s)
- Arashdeep Kaur
- Department of Microbiology, Panjab University, Sector-25, Chandigarh, 160014, India
| | - Sanjeev Kumar Soni
- Department of Microbiology, Panjab University, Sector-25, Chandigarh, 160014, India
| | - Shania Vij
- Department of Microbiology, Panjab University, Sector-25, Chandigarh, 160014, India
| | - Praveen Rishi
- Department of Microbiology, Panjab University, Sector-25, Chandigarh, 160014, India.
| |
Collapse
|
8
|
Mazurek-Popczyk J, Pisarska J, Bok E, Baldy-Chudzik K. Antibacterial Activity of Bacteriocinogenic Commensal Escherichia coli against Zoonotic Strains Resistant and Sensitive to Antibiotics. Antibiotics (Basel) 2020; 9:E411. [PMID: 32679778 PMCID: PMC7400030 DOI: 10.3390/antibiotics9070411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/23/2022] Open
Abstract
Antibiotic resistance concerns various areas with high consumption of antibiotics, including husbandry. Resistant strains are transmitted to humans from livestock and agricultural products via the food chain and may pose a health risk. The commensal microbiota protects against the invasion of environmental strains by secretion of bacteriocins, among other mechanisms. The present study aims to characterize the bactericidal potential of bacteriocinogenic Escherichia coli from healthy humans against multidrug-resistant and antibiotic-sensitive strains from pigs and cattle. Bacteriocin production was tested by the double-layer plate method, and bacteriocin genes were identified by the PCR method. At least one bacteriocinogenic E. coli was detected in the fecal samples of 55% of tested individuals, adults and children. Among all isolates (n = 210), 37.1% were bacteriocinogenic and contained genes of colicin (Col) Ib, ColE1, microcin (Mcc) H47, ColIa, ColM, MccV, ColK, ColB, and single ColE2 and ColE7. Twenty-five E. coli carrying various sets of bacteriocin genes were further characterized and tested for their activity against zoonotic strains (n = 60). Strains with ColE7 (88%), ColE1-ColIa-ColK-MccH47 (85%), MccH47-MccV (85%), ColE1-ColIa-ColM (82%), ColE1 (75%), ColM (67%), and ColK (65%) were most active against zoonotic strains. Statistically significant differences in activity toward antibiotic-resistant strains were shown by commensal E. coli carrying MccV, ColK-MccV, and ColIb-ColK. The study demonstrates that bacteriocinogenic commensal E. coli exerts antagonistic activity against zoonotic strains and may constitute a defense line against multidrug-resistant strains.
Collapse
Affiliation(s)
- Justyna Mazurek-Popczyk
- Department of Microbiology and Molecular Biology, Collegium Medicum, University of Zielona Góra, 65-417 Zielona Góra, Poland; (J.P.); (E.B.); (K.B.-C.)
| | | | | | | |
Collapse
|
9
|
Sharma G, Dang S, K A, Kalia M, Gabrani R. Synergistic antibacterial and anti-biofilm activity of nisin like bacteriocin with curcumin and cinnamaldehyde against ESBL and MBL producing clinical strains. BIOFOULING 2020; 36:710-724. [PMID: 32772715 DOI: 10.1080/08927014.2020.1804553] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 06/11/2023]
Abstract
Bacteriocins are small peptides that can inhibit the growth of a diverse range of microbes. There is a need to identify bacteriocins that are effective against biofilms of resistant clinical strains. The present study focussed on the efficacy of purified nisin like bacteriocin-GAM217 against extended spectrum β-lactamase (ESBL) and metallo-beta-lactamase (MBL) producing clinical strains. Bacteriocin-GAM217 when combined with curcumin and cinnamaldehyde, synergistically enhanced antibacterial activity against planktonic and biofilm cultures of Staphylococcus epidermidis and Escherichia coli. Bacteriocin-GAM217 and phytochemical combinations inhibited biofilm formation by >80%, and disrupted the biofilm for selected ESBL and MBL producing clinical strains. The anti-adhesion assay showed that these combinatorial compounds significantly lowered the attachment of bacteria to Vero cells and that they elicited membrane permeability and rapid killing as viewed by confocal microscopy. This study demonstrates that bacteriocin-GAM217 in combination with phytochemicals can be a potential anti-biofilm agent and thus has potential for biomedical applications.
Collapse
Affiliation(s)
- Garima Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Aruna K
- Department of Microbiology, Wilson College, Mumbai, India
| | | | - Reema Gabrani
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
10
|
Li J, Fernández-Millán P, Boix E. Synergism between Host Defence Peptides and Antibiotics Against Bacterial Infections. Curr Top Med Chem 2020; 20:1238-1263. [DOI: 10.2174/1568026620666200303122626] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 01/10/2023]
Abstract
Background:Antimicrobial resistance (AMR) to conventional antibiotics is becoming one of the main global health threats and novel alternative strategies are urging. Antimicrobial peptides (AMPs), once forgotten, are coming back into the scene as promising tools to overcome bacterial resistance. Recent findings have attracted attention to the potentiality of AMPs to work as antibiotic adjuvants.Methods:In this review, we have tried to collect the currently available information on the mechanism of action of AMPs in synergy with other antimicrobial agents. In particular, we have focused on the mechanisms of action that mediate the inhibition of the emergence of bacterial resistance by AMPs.Results and Conclusion:We find in the literature many examples where AMPs can significantly reduce the antibiotic effective concentration. Mainly, the peptides work at the bacterial cell wall and thereby facilitate the drug access to its intracellular target. Complementarily, AMPs can also contribute to permeate the exopolysaccharide layer of biofilm communities, or even prevent bacterial adhesion and biofilm growth. Secondly, we find other peptides that can directly block the emergence of bacterial resistance mechanisms or interfere with the community quorum-sensing systems. Interestingly, the effective peptide concentrations for adjuvant activity and inhibition of bacterial resistance are much lower than the required for direct antimicrobial action. Finally, many AMPs expressed by innate immune cells are endowed with immunomodulatory properties and can participate in the host response against infection. Recent studies in animal models confirm that AMPs work as adjuvants at non-toxic concentrations and can be safely administrated for novel combined chemotherapies.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Pablo Fernández-Millán
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| |
Collapse
|
11
|
Bartkiene E, Ruzauskas M, Bartkevics V, Pugajeva I, Zavistanaviciute P, Starkute V, Zokaityte E, Lele V, Dauksiene A, Grashorn M, Hoelzle LE, Mendybayeva A, Ryshyanova R, Gruzauskas R. Study of the antibiotic residues in poultry meat in some of the EU countries and selection of the best compositions of lactic acid bacteria and essential oils against Salmonella enterica. Poult Sci 2020; 99:4065-4076. [PMID: 32731994 PMCID: PMC7597929 DOI: 10.1016/j.psj.2020.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 04/27/2020] [Accepted: 05/01/2020] [Indexed: 11/18/2022] Open
Abstract
In this study, the presence of antibiotics (ANB) residues was evaluated in poultry meat purchased from German and Lithuanian markets. In addition, the antimicrobial activity of 13 lactic acid bacteria (LAB) strains, 2 essential oils (EO) (Thymus vulgaris and Origanum vulgare L.), and their compositions were tested for the purpose of inhibiting antibiotic-resistant Salmonella spp. ANB residues were found in 3 out of the 20 analyzed poultry meat samples: sample no. 8 contained enrofloxacin (0.46 μg/kg), sample no. 14 contained both enrofloxacin and doxycycline (0.05 and 16.8 μg/kg, respectively), and sample no. 18 contained enrofloxacin (2.06 μg/kg). The maximum residue limits (MRLs) for the sum of enrofloxacin and ciprofloxacin and for doxycycline in the poultry muscle are 100 μg/kg. Finally, none of the tested poultry meat samples exceeded the suggested MRLs; however, the issue of ANB residues still requires monitoring of the poultry industry in Germany, Poland, and Lithuania, despite the currently established low ANB concentrations. These findings can be explained by the increased use of alternatives to ANB in the poultry industry. Our results showed that an effective alternative to ANB, which can help to reduce the occurrence of antibiotic-resistant salmonella, is a composition containing 1.0% of thyme EO and the following LAB strains: Lactobacillus plantrum LUHS122, Enteroccocus pseudoavium LUHS242, Lactobacillus casei LUHS210, Lactobacillus paracasei LUHS244, Lactobacillus plantarum LUHS135, Lactobacillus coryniformins LUHS71, and Lactobacillus uvarum LUHS245, which can be recommended for poultry industry as components of feed or for the treatment of surfaces, to control the contamination with Salmonella strains. However, it should be mentioned that most of the tested LAB strains were inhibited by thyme EO at the concentrations of 0.5 and 1.0%, except for LUHS122, LUHS210, and LUHS245. Finally, it can be noted that the agents responsible for the inhibitory effect on Salmonella are not the viable LAB strains but rather their metabolites, and further studies are needed to identify which metabolites are the most important.
Collapse
Affiliation(s)
- Elena Bartkiene
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Food Safety and Quality, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania.
| | - Modestas Ruzauskas
- Institute of Microbiology and Virology, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Anatomy and Physiology, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania
| | - Vadims Bartkevics
- Institute of Food Safety, Animal Health and Environment BIOR, 1076 Riga, Latvia
| | - Iveta Pugajeva
- Institute of Food Safety, Animal Health and Environment BIOR, 1076 Riga, Latvia
| | - Paulina Zavistanaviciute
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Food Safety and Quality, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania
| | - Vytaute Starkute
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Food Safety and Quality, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania
| | - Egle Zokaityte
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Food Safety and Quality, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania
| | - Vita Lele
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Food Safety and Quality, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania
| | - Agila Dauksiene
- Institute of Animal Rearing Technologies, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania; Department of Anatomy and Physiology, Lithuanian University of Health Sciences, 47181, Kaunas, Lithuania
| | - Michael Grashorn
- Institute of Animal Science at University of Hohenheim, 70599 Stuttgart, Germany
| | - Ludwig E Hoelzle
- Institute of Animal Science at University of Hohenheim, 70599 Stuttgart, Germany
| | - Anara Mendybayeva
- Research Institute of Applied Biotechnology, Kostanay State University, 110000 Kostanay, Kazakhstan
| | - Raushan Ryshyanova
- Research Institute of Applied Biotechnology, Kostanay State University, 110000 Kostanay, Kazakhstan
| | - Romas Gruzauskas
- Department of Food Science and Technology, Kaunas University of Technology, 50254, Kaunas, Lithuania
| |
Collapse
|
12
|
Sharma P, Rashid M, Kaur S. Novel enterocin E20c purified from Enterococcus hirae 20c synergised with ß-lactams and ciprofloxacin against Salmonella enterica. Microb Cell Fact 2020; 19:98. [PMID: 32366243 PMCID: PMC7197179 DOI: 10.1186/s12934-020-01352-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
Background An increasing rate of antibiotic resistance among Gram-negative bacterial pathogens has created an urgent need to discover novel therapeutic agents to combat infectious diseases. Use of bacteriocins as therapeutic agents has immense potential due to their high potency and mode of action different from that of conventional antibiotics. Results In this study, a novel bacteriocin E20c of molecular weight 6.5 kDa was purified and characterized from the probiotic strain of Enterococcus hirae. E20c had bactericidal activities against several multidrug resistant (MDR) Gram-negative bacterial pathogens. Flow cytometry and scanning electron microscopy studies showed that it killed the Salmonella enterica cells by forming ion-permeable channels in the cell membrane leading to enhanced cell membrane permeability. Further, checkerboard titrations showed that E20c had synergistic interaction with antibiotics such as ampicillin, penicillin, ceftriaxone, and ciprofloxacin against a ciprofloxacin- and penicillin-resistant strain of S. enterica. Conclusion Thus, this study shows the broad spectrum antimicrobial activity of novel enterocin E20c against various MDR pathogens. Further, it highlights the importance of bacteriocins in lowering the minimum inhibitory concentrations of conventional antibiotics when used in combination.
Collapse
Affiliation(s)
- Preeti Sharma
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Muzamil Rashid
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Sukhraj Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, Punjab, India.
| |
Collapse
|
13
|
Nguyen T, Brody H, Lin GH, Rangé H, Kuraji R, Ye C, Kamarajan P, Radaic A, Gao L, Kapila Y. Probiotics, including nisin-based probiotics, improve clinical and microbial outcomes relevant to oral and systemic diseases. Periodontol 2000 2020; 82:173-185. [PMID: 31850634 DOI: 10.1111/prd.12324] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The effects of probiotic supplementation on systemic health and gastrointestinal diseases have been investigated in numerous studies. The aim of this review is to provide an overview of probiotics and their effects on periodontal health. Probiotics show beneficial effects as adjunctive therapeutics and as stand-alone agents in the treatment and prevention of gingivitis as well as specific clinical parameters of periodontitis. This review focuses on the clinical and microbiological aspects of probiotics in the context of health, gingivitis, and periodontitis. In addition, a special focus on nisin-producing probiotics and nisin itself showcase their significant potential for oral and systemic use.
Collapse
Affiliation(s)
- Trang Nguyen
- School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Hanna Brody
- School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Guo-Hao Lin
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Hélène Rangé
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA.,Department of Periodontology, UFR of Odontology, APHP, Rothschild Hospital, University of Paris Diderot, Paris, France.,Faculty of Dental Surgery, University of Paris Descartes, Montrouge, France
| | - Ryutaro Kuraji
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA.,Department of Life Science Dentistry, The Nippon Dental University, Tokyo, Japan.,Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Changchang Ye
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA.,State Key Laboratory of Oral Diseases, Department of Periodontology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pachiyappan Kamarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Allan Radaic
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA.,Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, The University of Campinas, Campinas, Sao Paulo, Brazil
| | - Li Gao
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA.,Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yvonne Kapila
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Alves FCB, Albano M, Andrade BFMT, Chechi JL, Pereira AFM, Furlanetto A, Rall VLM, Fernandes AAH, dos Santos LD, Barbosa LN, Fernandes Junior A. Comparative Proteomics of Methicillin-Resistant Staphylococcus aureus Subjected to Synergistic Effects of the Lantibiotic Nisin and Oxacillin. Microb Drug Resist 2020; 26:179-189. [DOI: 10.1089/mdr.2019.0038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Fernanda Cristina Bergamo Alves
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Mariana Albano
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | | | - Jéssica Luana Chechi
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Ana Flávia Marques Pereira
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Alessandra Furlanetto
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Vera Lúcia Mores Rall
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Ana Angélica Henrique Fernandes
- Department of Chemistry and Biochemistry, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| | - Lucilene Delazari dos Santos
- Graduate Program in Tropical Diseases, Botucatu Medical School (FMB), São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
- Center for the Study of Venom and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Lidiane Nunes Barbosa
- Graduate Program in Animal Sciences with Emphasis on Bioactive Products, Universidade Paranaense (UNIPAR), Umuarama, Brazil
| | - Ary Fernandes Junior
- Department of Microbiology and Immunology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
- Electronic Microscopy Center, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
15
|
Ducarmon QR, Zwittink RD, Hornung BVH, van Schaik W, Young VB, Kuijper EJ. Gut Microbiota and Colonization Resistance against Bacterial Enteric Infection. Microbiol Mol Biol Rev 2019; 83:e00007-19. [PMID: 31167904 PMCID: PMC6710460 DOI: 10.1128/mmbr.00007-19] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The gut microbiome is critical in providing resistance against colonization by exogenous microorganisms. The mechanisms via which the gut microbiota provide colonization resistance (CR) have not been fully elucidated, but they include secretion of antimicrobial products, nutrient competition, support of gut barrier integrity, and bacteriophage deployment. However, bacterial enteric infections are an important cause of disease globally, indicating that microbiota-mediated CR can be disturbed and become ineffective. Changes in microbiota composition, and potential subsequent disruption of CR, can be caused by various drugs, such as antibiotics, proton pump inhibitors, antidiabetics, and antipsychotics, thereby providing opportunities for exogenous pathogens to colonize the gut and ultimately cause infection. In addition, the most prevalent bacterial enteropathogens, including Clostridioides difficile, Salmonella enterica serovar Typhimurium, enterohemorrhagic Escherichia coli, Shigella flexneri, Campylobacter jejuni, Vibrio cholerae, Yersinia enterocolitica, and Listeria monocytogenes, can employ a wide array of mechanisms to overcome colonization resistance. This review aims to summarize current knowledge on how the gut microbiota can mediate colonization resistance against bacterial enteric infection and on how bacterial enteropathogens can overcome this resistance.
Collapse
Affiliation(s)
- Q R Ducarmon
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - R D Zwittink
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - B V H Hornung
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - W van Schaik
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - V B Young
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - E J Kuijper
- Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands
- Experimental Bacteriology, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
- Clinical Microbiology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
- Netherlands Donor Feces Bank, Leiden, Netherlands
| |
Collapse
|
16
|
Vieco-Saiz N, Belguesmia Y, Raspoet R, Auclair E, Gancel F, Kempf I, Drider D. Benefits and Inputs From Lactic Acid Bacteria and Their Bacteriocins as Alternatives to Antibiotic Growth Promoters During Food-Animal Production. Front Microbiol 2019; 10:57. [PMID: 30804896 PMCID: PMC6378274 DOI: 10.3389/fmicb.2019.00057] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/14/2019] [Indexed: 12/27/2022] Open
Abstract
Resistance to antibiotics is escalating and threatening humans and animals worldwide. Different countries have legislated or promoted the ban of antibiotics as growth promoters in livestock and aquaculture to reduce this phenomenon. Therefore, to improve animal growth and reproduction performance and to control multiple bacterial infections, there is a potential to use probiotics as non-antibiotic growth promoters. Lactic acid bacteria (LAB) offer various advantages as potential probiotics and can be considered as alternatives to antibiotics during food-animal production. LAB are safe microorganisms with abilities to produce different inhibitory compounds such as bacteriocins, organic acids as lactic acid, hydrogen peroxide, diacetyl, and carbon dioxide. LAB can inhibit harmful microorganisms with their arsenal, or through competitive exclusion mechanism based on competition for binding sites and nutrients. LAB endowed with specific enzymatic functions (amylase, protease…) can improve nutrients acquisition as well as animal immune system stimulation. This review aimed at underlining the benefits and inputs from LAB as potential alternatives to antibiotics in poultry, pigs, ruminants, and aquaculture production.
Collapse
Affiliation(s)
- Nuria Vieco-Saiz
- EA7394-ICV, Institut Charles Viollette, Université de Lille, Villeneuve-d’Ascq, France
- Phileo Lesaffre Animal Care, Marcq-en-Barœul, France
| | - Yanath Belguesmia
- EA7394-ICV, Institut Charles Viollette, Université de Lille, Villeneuve-d’Ascq, France
| | - Ruth Raspoet
- Phileo Lesaffre Animal Care, Marcq-en-Barœul, France
| | - Eric Auclair
- Phileo Lesaffre Animal Care, Marcq-en-Barœul, France
| | - Frédérique Gancel
- EA7394-ICV, Institut Charles Viollette, Université de Lille, Villeneuve-d’Ascq, France
| | - Isabelle Kempf
- Laboratoire de Ploufragan-Plouzané-Niort, Agence Nationale de Sécurité Sanitaire de l’Alimentation, de l’Environnement et du Travail (ANSES), Ploufragan, France
- Université Bretagne Loire, Rennes, France
| | - Djamel Drider
- EA7394-ICV, Institut Charles Viollette, Université de Lille, Villeneuve-d’Ascq, France
| |
Collapse
|
17
|
Rishi P, Vashist T, Sharma A, Kaur A, Kaur A, Kaur N, Kaur IP, Tewari R. Efficacy of designer K11 antimicrobial peptide (a hybrid of melittin, cecropin A1 and magainin 2) against Acinetobacter baumannii-infected wounds. Pathog Dis 2018; 76:5089976. [PMID: 30184071 DOI: 10.1093/femspd/fty072] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/01/2018] [Indexed: 01/04/2025] Open
Abstract
Due to emergence of multidrug resistance in pathogens, the attention of the scientific community is now directed towards strengthening the reservoir of antimicrobial compounds. Prior to in vivo studies, the interaction and penetration of a hybrid peptide K11 in bacterial cells using confocal microscopy was assessed which was observed as early as 10 min after incubation with the peptide. Cell lysis along with leakage of cytoplasmic content was confirmed by electron microscopy. To evaluate the in vivo performance of the peptide, it was contained in carbopol hydrogel. Efficacy of the hydrogel formulation was then evaluated against Acinetobacter baumannii-infected wounds using a murine excision model. Treatment resulted in restoration of body weight, complete clearance of infection from the wound by day 7 and 99% wound enclosure by day 21, in contrast to the persistence of infection and 70% wound enclosure in the infected group. Further, this treatment resulted in a 2.6-fold decrease in the levels of malondialdehyde along with a 4.5-fold increase in the levels of catalase on day 3. Appearance of normal histo-architecture was observed in the treatment group. Based on these results, the peptide hydrogel can be exploited in future as one of the strategies for developing a topical anti-infective therapeutic agent.
Collapse
Affiliation(s)
- Praveen Rishi
- Department of Microbiology, Basic Medical Sciences Block I, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
| | - Tanvi Vashist
- Department of Microbiology, Basic Medical Sciences Block I, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
| | - Avantika Sharma
- Department of Microbial Biotechnology, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
| | - Amrita Kaur
- Department of Microbiology, Basic Medical Sciences Block I, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
| | - Arashdeep Kaur
- Department of Microbiology, Basic Medical Sciences Block I, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
| | - Navneet Kaur
- Department of Microbiology, Basic Medical Sciences Block I, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
- CSIR-Institute of Microbial Technology, Sector-39 A, Chandigarh-160036, India
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh-160014, India
| | - Rupinder Tewari
- Department of Microbial Biotechnology, South Campus, Sector 25, Panjab University, Chandigarh-160014, India
| |
Collapse
|
18
|
Lewies A, Du Plessis LH, Wentzel JF. Antimicrobial Peptides: the Achilles’ Heel of Antibiotic Resistance? Probiotics Antimicrob Proteins 2018; 11:370-381. [DOI: 10.1007/s12602-018-9465-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
19
|
Rishi P, Vij S, Maurya IK, Kaur UJ, Bharati S, Tewari R. Peptides as adjuvants for ampicillin and oxacillin against methicillin-resistant Staphylococcus aureus (MRSA). Microb Pathog 2018; 124:11-20. [PMID: 30118800 DOI: 10.1016/j.micpath.2018.08.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/12/2018] [Accepted: 08/13/2018] [Indexed: 01/15/2023]
Abstract
Fast emerging antibiotic resistance in pathogens requires special attention for strengthening the reservoir of antimicrobial compounds. In view of this, several peptides with known antimicrobial activities have been reported to enhance the efficacy of antibiotics against multidrug resistant (MDR) pathogens. In the present study, potential of peptides having distinct mechanism of action, if any, was evaluated to improve the efficacy of conventional antibiotics against methicillin-resistant S. aureus (MRSA). After primary screening of six peptides, two peptides namely T3 and T4 showing very high minimum inhibitory concentrations (MICs) were selected to assess their role in altering the MICs of antibiotics to which the pathogen was resistant. In the presence of the peptides, the MICs of the antibiotics were found to be reduced as per the fractional inhibitory concentration indices (FICI) and time kill assay. These observations prompted us to look for their mechanism of action. The effect of peptides on the morphology of pathogen by field emission scanning electron microscopy (FE-SEM) revealed no damage to the cells at the sub-inhibitory concentrations of the peptide which correlated well with the higher MIC of the peptide, indicating no direct impact on the pathogen. However, dielectric spectroscopy, confocal microscopy and flow cytometry confirmed the interaction and localization of peptides with the bacterial membrane. The peptides were also found to inhibit efflux of ethidium bromide which is the substrate for many proteins involved in efflux system. Therefore, it is speculated that the peptides after interacting with the membrane of the pathogen might have resulted in the inhibition of the efflux of antibiotics thereby reducing their effective concentrations. The study thus suggests that peptides with no antimicrobial activity of their own, can also enhance the efficacy of the antibiotics by interacting with the pathogen thereby, acting as adjuvants for the antibiotics.
Collapse
Affiliation(s)
- Praveen Rishi
- Department of Microbiology, Basic Medical Sciences Block-I, Panjab University, Chandigarh, India.
| | - Shania Vij
- Department of Microbiology, Basic Medical Sciences Block-I, Panjab University, Chandigarh, India
| | | | - Ujjwal Jit Kaur
- Department of Microbiology, Basic Medical Sciences Block-I, Panjab University, Chandigarh, India
| | - Sanjay Bharati
- Department of Nuclear Medicine, School of Allied Health Sciences, Manipal University, Manipal, Karnataka, India
| | - Rupinder Tewari
- Department of Microbial Biotechnology, Panjab University, Chandigarh, India
| |
Collapse
|
20
|
Bhattacharya D, Ghosh D, Bhattacharya S, Sarkar S, Karmakar P, Koley H, Gachhui R. Antibacterial activity of polyphenolic fraction of Kombucha against Vibrio cholerae: targeting cell membrane. Lett Appl Microbiol 2018; 66:145-152. [PMID: 29193174 DOI: 10.1111/lam.12829] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 11/28/2022]
Abstract
The present study was undertaken to determine the mechanism of antibacterial activity of a polyphenolic fraction, composed of mainly catechin and isorhamnetin, previously isolated from Kombucha, a 14-day fermented beverage of sugared black tea, against the enteropathogen Vibrio cholerae N16961. Bacterial growth was found to be seriously impaired by the polyphenolic fraction in a dose-dependent manner. Scanning Electron Microscopy demonstrated morphological alterations in bacterial cells when exposed to the polyphenolic fraction in a concentration-dependent manner. Permeabilization assays confirmed that the fraction disrupted bacterial membrane integrity in both time- and dose-dependent manners, which were proportional to the production of intracellular reactive oxygen species (ROS). Furthermore, each of the polyphenols catechin and isorhamnetin showed the ability to permeate bacterial cell membranes by generating oxidative stress, thereby suggesting their role in the antibacterial potential of Kombucha. Thus, the basic mechanism of antibacterial activity of the Kombucha polyphenolic fraction against V. cholerae involved bacterial membrane permeabilization and morphological changes, which might be due to the generation of intracellular ROS. To the best of our knowledge, this is the first report on the investigation of antibacterial mechanism of Kombucha, which is mostly attributed to its polyphenolic content. SIGNIFICANCE AND IMPACT OF THE STUDY The emergence of multidrug-resistant Vibrio cholerae strains has hindered an efficient anti-Vibrio therapy. This study has demonstrated the membrane damage-mediated antibacterial mechanism of Kombucha, a popular fermented beverage of sugared tea, which is mostly attributed to its polyphenolic content. This study also implies the exploitation of Kombucha as a potential new source of bioactive polyphenols against V. cholerae.
Collapse
Affiliation(s)
- D Bhattacharya
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata, India
| | - D Ghosh
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata, India
| | - S Bhattacharya
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata, India
| | - S Sarkar
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata, India
| | - P Karmakar
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata, India
| | - H Koley
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases (NICED), Beliaghata, Kolkata, India
| | - R Gachhui
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata, India
| |
Collapse
|
21
|
Rishi P, Bhagat NR, Thakur R, Pathania P. Tackling Salmonella Persister Cells by Antibiotic-Nisin Combination via Mannitol. Indian J Microbiol 2018; 58:239-243. [PMID: 29651185 DOI: 10.1007/s12088-018-0713-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
Bacterial persisters (defined as dormant, non-dividing cells with globally reduced metabolism) are the major cause of recurrent infections. As they neither grow nor die in presence of antibiotics, it is difficult to eradicate these cells using antibiotics, even at higher concentrations. Reports of metabolites (which help in waking up of these inactive cells) enabled eradication of bacterial persistence by aminoglycosides, suggest the new potential strategy to improve antibiotic therapy. Here we propose, mannitol enabled elimination of Salmonella persister cells by the nisin-antibiotic combination. For this, persister cells were developed and characterized for their typical properties such as non-replicative state and metabolic dormancy. Different carbon sources viz. glucose, glycerol, and mannitol were used, each as an adjunct to ampicillin for the eradication of persister cells. The maximum (but not complete) killing was observed with mannitol-ampicillin, out of all the combinations used. However, significant elimination (about 78%) could be observed, when nisin (an antimicrobial peptide) was used with ampicillin in presence of mannitol, which might have mediated the transfer of antibiotic-nisin combination at the same time when the cells tried to grab the carbon molecule. Further, the effectiveness of the trio was confirmed by flow cytometry. Overall, our findings highlight the potential of this trio-combination for developing it as an option for tackling Salmonella persister cells.
Collapse
Affiliation(s)
- Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
| | - Neha Rani Bhagat
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
| | - Reena Thakur
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
| | - Preeti Pathania
- Department of Microbiology, Panjab University, Chandigarh, 160014 India
| |
Collapse
|
22
|
Bacteriocin-like inhibitory substance (BLIS) activity of Streptococcus macedonicus MBF10-2 and its synergistic action in combination with antibiotics. ASIAN PAC J TROP MED 2017; 10:1140-1145. [DOI: 10.1016/j.apjtm.2017.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 08/18/2017] [Accepted: 09/07/2017] [Indexed: 11/19/2022] Open
|
23
|
Mathur H, Field D, Rea MC, Cotter PD, Hill C, Ross RP. Bacteriocin-Antimicrobial Synergy: A Medical and Food Perspective. Front Microbiol 2017; 8:1205. [PMID: 28706513 PMCID: PMC5489601 DOI: 10.3389/fmicb.2017.01205] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/14/2017] [Indexed: 12/18/2022] Open
Abstract
The continuing emergence of multi-drug resistant pathogens has sparked an interest in seeking alternative therapeutic options. Antimicrobial combinatorial therapy is one such avenue. A number of studies have been conducted, involving combinations of bacteriocins with other antimicrobials, to circumvent the development of antimicrobial resistance and/or increase antimicrobial potency. Such bacteriocin-antimicrobial combinations could have tremendous value, in terms of reducing the likelihood of resistance development due to the involvement of two distinct mechanisms of antimicrobial action. Furthermore, antimicrobial synergistic interactions may also have potential financial implications in terms of decreasing the costs of treatment by reducing the concentration of an expensive antimicrobial and utilizing it in combination with an inexpensive one. In addition, combinatorial therapies with bacteriocins can broaden antimicrobial spectra and/or result in a reduction in the concentration of an antibiotic required for effective treatments to the extent that potentially toxic or adverse side effects can be reduced or eliminated. Here, we review studies in which bacteriocins were found to be effective in combination with other antimicrobials, with a view to targeting clinical and/or food-borne pathogens. Furthermore, we discuss some of the bottlenecks which are currently hindering the development of bacteriocins as viable therapeutic options, as well as addressing the need to exercise caution when attempting to predict clinical outcomes of bacteriocin-antimicrobial combinations.
Collapse
Affiliation(s)
- Harsh Mathur
- Teagasc Food Research Centre, MooreparkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Des Field
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| | - Mary C Rea
- Teagasc Food Research Centre, MooreparkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Centre, MooreparkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Colin Hill
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| | - R Paul Ross
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| |
Collapse
|
24
|
Singh AP, Prabha V, Rishi P. Synergism in dual functionality of cryptdin-2 in conjunction with antibiotics against Salmonella. Indian J Med Res 2017; 144:761-770. [PMID: 28361830 PMCID: PMC5393088 DOI: 10.4103/ijmr.ijmr_850_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND & OBJECTIVES The emergence of multidrug-resistant Salmonella over the last two decades poses a major health risk. In this context, antimicrobial peptides have found a strategic place in the therapeutic armamentarium. Previously, we found that cryptdin-2 has the potential to augment the activity of conventional second- and third-generation anti-Salmonella antibiotics as evident by in vitro assays. In continuation to this, the present study was designed to evaluate the in vivo synergistic effects, if any, of cryptdin-2 in combination with ciprofloxacin and ceftriaxone against murine salmonellosis. METHODS Scanning electron microscopy (SEM) studies along with in vivo synergistic studies were performed using cryptdin- 2 and antibiotic combinations. In addition, peroxidative liver damage, levels of nitric oxide (NO) and antioxidant enzymes along with tumour necrosis factor-alpha (TNF-α) levels were also measured. RESULTS The SEM results revealed marked changes on the outer membrane of the bacterial cells treated with various combinations. Both the tested combinations demonstrated synergistic in vivo potency against S. Typhimurium as evident by reduction in the number of Salmonellae in the liver, spleen and intestine. Analysis of peroxidative liver damage, levels of NO and antioxidant enzymes along with TNF-α and nuclear factor-kappa B levels revealed that the tested combinations restored their levels to near normal. The most potent combination was found to be that of cryptdin-2 and ciprofloxacin in terms of direct killing and immunomodulatory potential. INTERPRETATION & CONCLUSIONS These findings suggest that cryptdin-2 may act in conjunction with conventional antibiotics indicating the possibility of developing these combinations as additional therapeutic agents to combat Salmonella infections.
Collapse
Affiliation(s)
- Aman Preet Singh
- Department of Microbiology, Basic Medical Sciences Block, Panjab University, Chandigarh, India
| | - Vijay Prabha
- Department of Microbiology, Basic Medical Sciences Block, Panjab University, Chandigarh, India
| | - Praveen Rishi
- Department of Microbiology, Basic Medical Sciences Block, Panjab University, Chandigarh, India
| |
Collapse
|
25
|
Ongey EL, Neubauer P. Lanthipeptides: chemical synthesis versus in vivo biosynthesis as tools for pharmaceutical production. Microb Cell Fact 2016; 15:97. [PMID: 27267232 PMCID: PMC4897893 DOI: 10.1186/s12934-016-0502-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/01/2016] [Indexed: 01/15/2023] Open
Abstract
Lanthipeptides (also called lantibiotics for those with antibacterial activities) are ribosomally synthesized post-translationally modified peptides having thioether cross-linked amino acids, lanthionines, as a structural element. Lanthipeptides have conceivable potentials to be used as therapeutics, however, the lack of stable, high-yield, well-characterized processes for their sustainable production limit their availability for clinical studies and further pharmaceutical commercialization. Though many reviews have discussed the various techniques that are currently employed to produce lanthipeptides, a direct comparison between these methods to assess industrial applicability has not yet been described. In this review we provide a synoptic comparison of research efforts on total synthesis and in vivo biosynthesis aimed at fostering lanthipeptides production. We further examine current applications and propose measures to enhance product yields. Owing to their elaborate chemical structures, chemical synthesis of these biomolecules is economically less feasible for large-scale applications, and hence biological production seems to be the only realistic alternative.
Collapse
Affiliation(s)
- Elvis Legala Ongey
- Chair of Bioprocess Engineering, Department of Biotechnology, Technische Universität Berlin, Ackerstraße 76, ACK24, 13355, Berlin, Germany.
| | - Peter Neubauer
- Chair of Bioprocess Engineering, Department of Biotechnology, Technische Universität Berlin, Ackerstraße 76, ACK24, 13355, Berlin, Germany
| |
Collapse
|
26
|
Al Atya AK, Belguesmia Y, Chataigne G, Ravallec R, Vachée A, Szunerits S, Boukherroub R, Drider D. Anti-MRSA Activities of Enterocins DD28 and DD93 and Evidences on Their Role in the Inhibition of Biofilm Formation. Front Microbiol 2016; 7:817. [PMID: 27303396 PMCID: PMC4886693 DOI: 10.3389/fmicb.2016.00817] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 05/13/2016] [Indexed: 01/04/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has become a worrisome superbug. This work aimed at studying the effects of two class IIb bacteriocins, enterocins DD28 and DD93 as anti-MRSA agents. Thus, these bacteriocins were purified, from the cultures supernatants of Enterococcus faecalis 28 and 93, using a simplified purification procedure consisting in a cation exchange chromatography and a reversed-phase high-performance liquid chromatography. The anti-Staphylococcal activity was shown in vitro by the assessment of the minimal inhibitory concentration (MIC), followed by a checkerboard and time-kill kinetics experiments. The data unveiled a clear synergistic effect of enterocins DD28 and DD93 in combination with erythromycin or kanamycin against the clinical MRSA-S1 strain. Besides, these combinations impeded as well the MRSA-S1 clinical strain to setup biofilms on stainless steel and glace devices.
Collapse
Affiliation(s)
- Ahmed K Al Atya
- Université de Lille 1 Sciences et Technologies - Institut Charles Viollette Lille, France
| | - Yanath Belguesmia
- Université de Lille 1 Sciences et Technologies - Institut Charles Viollette Lille, France
| | - Gabrielle Chataigne
- Université de Lille 1 Sciences et Technologies - Institut Charles Viollette Lille, France
| | - Rozenn Ravallec
- Université de Lille 1 Sciences et Technologies - Institut Charles Viollette Lille, France
| | - Anne Vachée
- Hôpital Victor Provo de Roubaix Roubaix, France
| | - Sabine Szunerits
- Institut d'Electronique, de Microélectronique et de Nanotechnologie, UMR CNRS 8520, Université Lille 1 Lille, France
| | - Rabah Boukherroub
- Institut d'Electronique, de Microélectronique et de Nanotechnologie, UMR CNRS 8520, Université Lille 1 Lille, France
| | - Djamel Drider
- Université de Lille 1 Sciences et Technologies - Institut Charles Viollette Lille, France
| |
Collapse
|
27
|
Cavera VL, Arthur TD, Kashtanov D, Chikindas ML. Bacteriocins and their position in the next wave of conventional antibiotics. Int J Antimicrob Agents 2015; 46:494-501. [PMID: 26341839 DOI: 10.1016/j.ijantimicag.2015.07.011] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/10/2015] [Accepted: 07/15/2015] [Indexed: 12/13/2022]
Abstract
Micro-organisms are capable of producing a range of defence mechanisms, including antibiotics, bacteriocins, lytic agents, protein exotoxins, etc. Such mechanisms have been identified in nearly 99% of studied bacteria. The multiplicity and diversity of bacteriocins and the resultant effects of their interactions with targeted bacteria on microbial ecology has been thoroughly studied and remains an area of investigation attracting many researchers. However, the incorporation of bacteriocins into drug delivery systems used in conjunction with, or as potential alternatives to, conventional antibiotics is only a recent, although rapidly expanding, field. The extensive array of bacteriocins positions them as one of the most promising options in the next wave of antibiotics. The goal of this review was to explore bacteriocins as novel antimicrobials, alone and in combination with established antibiotics, and thus position them as a potential tool for addressing the current antibiotic crisis.
Collapse
Affiliation(s)
- Veronica L Cavera
- Department of Biochemistry and Microbiology, Rutgers State University, 76 Lipman Drive, New Brunswick, NJ 08901, USA
| | - Timothy D Arthur
- Department of Biochemistry and Microbiology, Rutgers State University, 76 Lipman Drive, New Brunswick, NJ 08901, USA
| | - Dimitri Kashtanov
- School of Environmental and Biological Sciences, Rutgers State University, 65 Dudley Road, New Brunswick, NJ 08901, USA
| | - Michael L Chikindas
- School of Environmental and Biological Sciences, Rutgers State University, 65 Dudley Road, New Brunswick, NJ 08901, USA.
| |
Collapse
|
28
|
Wang G, Mishra B, Lau K, Lushnikova T, Golla R, Wang X. Antimicrobial peptides in 2014. Pharmaceuticals (Basel) 2015; 8:123-50. [PMID: 25806720 PMCID: PMC4381204 DOI: 10.3390/ph8010123] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 12/13/2022] Open
Abstract
This article highlights new members, novel mechanisms of action, new functions, and interesting applications of antimicrobial peptides reported in 2014. As of December 2014, over 100 new peptides were registered into the Antimicrobial Peptide Database, increasing the total number of entries to 2493. Unique antimicrobial peptides have been identified from marine bacteria, fungi, and plants. Environmental conditions clearly influence peptide activity or function. Human α-defensin HD-6 is only antimicrobial under reduced conditions. The pH-dependent oligomerization of human cathelicidin LL-37 is linked to double-stranded RNA delivery to endosomes, where the acidic pH triggers the dissociation of the peptide aggregate to release its cargo. Proline-rich peptides, previously known to bind to heat shock proteins, are shown to inhibit protein synthesis. A model antimicrobial peptide is demonstrated to have multiple hits on bacteria, including surface protein delocalization. While cell surface modification to decrease cationic peptide binding is a recognized resistance mechanism for pathogenic bacteria, it is also used as a survival strategy for commensal bacteria. The year 2014 also witnessed continued efforts in exploiting potential applications of antimicrobial peptides. We highlight 3D structure-based design of peptide antimicrobials and vaccines, surface coating, delivery systems, and microbial detection devices involving antimicrobial peptides. The 2014 results also support that combination therapy is preferred over monotherapy in treating biofilms.
Collapse
Affiliation(s)
- Guangshun Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA.
| | - Biswajit Mishra
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Kyle Lau
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Tamara Lushnikova
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Radha Golla
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
| | - Xiuqing Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE 68198-6495, USA
- Institute of Clinical Laboratory, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|